首页 > 最新文献

Journal of Biochemical and Molecular Toxicology最新文献

英文 中文
Estrogen Promotes the Proliferation and Migration of Endometrial Cancer Through the GPER-Mediated NOTCH Pathway
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-29 DOI: 10.1002/jbt.70129
Meng Qi, Yuxi Jin, Lulu Si, Hanlin Fu, Xiaojing Shi, Yana Liu, Yifan Wang, Ruixia Guo

This study aims to investigate the expression of GPER in EC, assess the impact of estrogen on the proliferation and migration of EC via GPER, and examine the potential role of GPER in mediating the NOTCH pathway to influence EC proliferation and migration. The expression of GPER and its correlation with clinicopathological features were investigated using clinical data. Cell proliferation was assessed through MTT and EdU assays, while cell migration ability was evaluated using wound healing and transwell assays. Western blot analysis was conducted to detect proteins associated with the GPER and NOTCH signaling pathways. Additionally, xenograft tumor models were established to investigate the potential role of estrogen in mediating the NOTCH pathway via GPER. The results demonstrated a significant upregulation of GPER expression in EC, which was associated with clinical stage and metastasis. In vitro experiments provided evidence that estrogen promotes EC cell proliferation and metastasis by enhancing the expression levels of GPER, Notch1, and Hes-1 proteins. Conversely, knocking down or suppressing GPER effectively reverses these effects. Furthermore, treatment with JAG-1, an agonist for the NOTCH pathway, counteracts si-GPER's inhibitory impact on both proliferation and migration abilities of EC cells while increasing Notch1 and Hes-1 protein expression levels; however, it does not alter GPER expression. In vivo experiments have substantiated that estrogen facilitates EC proliferation via the GPER-mediated NOTCH pathway.

{"title":"Estrogen Promotes the Proliferation and Migration of Endometrial Cancer Through the GPER-Mediated NOTCH Pathway","authors":"Meng Qi,&nbsp;Yuxi Jin,&nbsp;Lulu Si,&nbsp;Hanlin Fu,&nbsp;Xiaojing Shi,&nbsp;Yana Liu,&nbsp;Yifan Wang,&nbsp;Ruixia Guo","doi":"10.1002/jbt.70129","DOIUrl":"10.1002/jbt.70129","url":null,"abstract":"<p>This study aims to investigate the expression of GPER in EC, assess the impact of estrogen on the proliferation and migration of EC via GPER, and examine the potential role of GPER in mediating the NOTCH pathway to influence EC proliferation and migration. The expression of GPER and its correlation with clinicopathological features were investigated using clinical data. Cell proliferation was assessed through MTT and EdU assays, while cell migration ability was evaluated using wound healing and transwell assays. Western blot analysis was conducted to detect proteins associated with the GPER and NOTCH signaling pathways. Additionally, xenograft tumor models were established to investigate the potential role of estrogen in mediating the NOTCH pathway via GPER. The results demonstrated a significant upregulation of GPER expression in EC, which was associated with clinical stage and metastasis. In vitro experiments provided evidence that estrogen promotes EC cell proliferation and metastasis by enhancing the expression levels of GPER, Notch1, and Hes-1 proteins. Conversely, knocking down or suppressing GPER effectively reverses these effects. Furthermore, treatment with JAG-1, an agonist for the NOTCH pathway, counteracts si-GPER's inhibitory impact on both proliferation and migration abilities of EC cells while increasing Notch1 and Hes-1 protein expression levels; however, it does not alter GPER expression. In vivo experiments have substantiated that estrogen facilitates EC proliferation via the GPER-mediated NOTCH pathway.</p>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 2","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11775877/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143059116","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploring the Role of Epithelial–Mesenchymal Transition During Colorectal Cancer Peritoneal Metastasis: Update on Their Mechanisms
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-27 DOI: 10.1002/jbt.70166
Chenyan Long, Xiang Li, Jungang Liu, Xianwei Mo, Huage Zhong, Weizhong Tang, Junfeng Yu

Colorectal cancer is a common malignant tumor worldwide. The prognosis of patients with colorectal cancer peritoneal metastasis is very poor. The study of the specific mechanisms of colorectal cancer peritoneal metastasis plays an important role in the treatment of patients with this disease. The mechanisms of colorectal cancer peritoneal metastasis are mainly pathological and biological. Biologically, the epithelial–mesenchymal transition process is an important precursor to tumor cell metastasis. Therefore, it is necessary to study the mechanisms of colorectal cancer peritoneal metastasis, especially the epithelial–mesenchymal transition, to identify new methods for the prevention and treatment of colorectal cancer peritoneal cancer, reduce the incidence of colorectal cancer peritoneal metastasis, and improve patient prognosis.

大肠癌是全球常见的恶性肿瘤。大肠癌腹膜转移患者的预后很差。研究大肠癌腹膜转移的具体机制对治疗该病患者具有重要作用。大肠癌腹膜转移的机制主要有病理学和生物学两方面。在生物学上,上皮-间质转化过程是肿瘤细胞转移的重要前体。因此,有必要对结直肠癌腹膜转移的机制,特别是上皮-间质转化机制进行研究,以找出防治结直肠癌腹膜癌的新方法,降低结直肠癌腹膜转移的发病率,改善患者预后。
{"title":"Exploring the Role of Epithelial–Mesenchymal Transition During Colorectal Cancer Peritoneal Metastasis: Update on Their Mechanisms","authors":"Chenyan Long,&nbsp;Xiang Li,&nbsp;Jungang Liu,&nbsp;Xianwei Mo,&nbsp;Huage Zhong,&nbsp;Weizhong Tang,&nbsp;Junfeng Yu","doi":"10.1002/jbt.70166","DOIUrl":"10.1002/jbt.70166","url":null,"abstract":"<div>\u0000 \u0000 <p>Colorectal cancer is a common malignant tumor worldwide. The prognosis of patients with colorectal cancer peritoneal metastasis is very poor. The study of the specific mechanisms of colorectal cancer peritoneal metastasis plays an important role in the treatment of patients with this disease. The mechanisms of colorectal cancer peritoneal metastasis are mainly pathological and biological. Biologically, the epithelial–mesenchymal transition process is an important precursor to tumor cell metastasis. Therefore, it is necessary to study the mechanisms of colorectal cancer peritoneal metastasis, especially the epithelial–mesenchymal transition, to identify new methods for the prevention and treatment of colorectal cancer peritoneal cancer, reduce the incidence of colorectal cancer peritoneal metastasis, and improve patient prognosis.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 2","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143052633","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unlocking the Potential of miRNAs in Sepsis Diagnosis and Prognosis: From Pathophysiology to Precision Medicine 发掘 miRNA 在败血症诊断和预后中的潜力:从病理生理学到精准医学。
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-27 DOI: 10.1002/jbt.70156
Emad Gamil Khidr, Gharieb S. El-Sayyad, Ahmed I. Abulsoud, Nehal I. Rizk, Mohamed Bakr Zaki, Ahmed Amr Raouf, Mahmoud A. Elrebehy, Manal M. M. Abdel Hady, Mohammed S. Elballal, Osama A. Mohammed, Mustafa Ahmed Abdel-Reheim, Walaa A. El-Dakroury, Sherif S. Abdel Mageed, Tohada M. AL-Noshokaty, Ahmed S. Doghish

The clinical syndrome appears as a dysregulated host response to infection that results in life-threatening organ dysfunction known as Sepsis. Sepsis is a serious public health concern where for every five deaths in ICU there is one patient who dies with sepsis worldwide. Sepsis is featured as unbalanced inflammation and immunosuppression which is sustained and profound, increasing patient susceptibility to secondary infections and mortality. microRNAs (miRNAs) play a central role in the control of many biological processes, and the deregulation of their expression has been linked to the development of oncological, cardiovascular, neurodegenerative, and metabolic diseases. In this review, we discuss the role of miRNAs in sepsis pathophysiology. Overall, miRNAs are seen as promising biomarkers, and it has been proposed to develop miRNA-based diagnosis and therapies for sepsis. Yet, the picture is not so straightforward because of miRNAs’ versatile and dynamic features. More research is needed to clarify the expression and role of miRNAs in sepsis and promote the use of miRNAs for sepsis management. This study provides an extensive, current, and thorough analysis of the involvement of miRNAs in sepsis. Its purpose is to encourage future research in this area, as tiny miRNAs have the potential to be used for rapid diagnosis, prognosis, and treatment of sepsis.

临床综合征表现为宿主对感染的反应失调,导致危及生命的器官功能障碍,即败血症。败血症是一个严重的公共卫生问题,在全球范围内,重症监护病房每死亡五人,就有一人死于败血症。脓毒症的特点是不平衡的炎症和免疫抑制,这种不平衡的炎症和免疫抑制是持续的、严重的,增加了患者继发感染的易感性和死亡率。microRNAs(miRNAs)在许多生物过程的控制中发挥着核心作用,其表达的失调与肿瘤、心血管、神经退行性疾病和代谢性疾病的发展有关。在这篇综述中,我们将讨论 miRNA 在败血症病理生理学中的作用。总体而言,miRNAs 被认为是有前景的生物标志物,有人建议开发基于 miRNA 的败血症诊断和疗法。然而,由于 miRNA 具有多变和动态的特点,情况并非如此简单。要明确 miRNA 在败血症中的表达和作用,促进 miRNA 在败血症治疗中的应用,还需要更多的研究。本研究对 miRNAs 在败血症中的参与进行了广泛、及时和深入的分析。其目的是鼓励今后在这一领域的研究,因为微小的 miRNA 有可能用于败血症的快速诊断、预后和治疗。
{"title":"Unlocking the Potential of miRNAs in Sepsis Diagnosis and Prognosis: From Pathophysiology to Precision Medicine","authors":"Emad Gamil Khidr,&nbsp;Gharieb S. El-Sayyad,&nbsp;Ahmed I. Abulsoud,&nbsp;Nehal I. Rizk,&nbsp;Mohamed Bakr Zaki,&nbsp;Ahmed Amr Raouf,&nbsp;Mahmoud A. Elrebehy,&nbsp;Manal M. M. Abdel Hady,&nbsp;Mohammed S. Elballal,&nbsp;Osama A. Mohammed,&nbsp;Mustafa Ahmed Abdel-Reheim,&nbsp;Walaa A. El-Dakroury,&nbsp;Sherif S. Abdel Mageed,&nbsp;Tohada M. AL-Noshokaty,&nbsp;Ahmed S. Doghish","doi":"10.1002/jbt.70156","DOIUrl":"10.1002/jbt.70156","url":null,"abstract":"<div>\u0000 \u0000 <p>The clinical syndrome appears as a dysregulated host response to infection that results in life-threatening organ dysfunction known as Sepsis. Sepsis is a serious public health concern where for every five deaths in ICU there is one patient who dies with sepsis worldwide. Sepsis is featured as unbalanced inflammation and immunosuppression which is sustained and profound, increasing patient susceptibility to secondary infections and mortality. microRNAs (miRNAs) play a central role in the control of many biological processes, and the deregulation of their expression has been linked to the development of oncological, cardiovascular, neurodegenerative, and metabolic diseases. In this review, we discuss the role of miRNAs in sepsis pathophysiology. Overall, miRNAs are seen as promising biomarkers, and it has been proposed to develop miRNA-based diagnosis and therapies for sepsis. Yet, the picture is not so straightforward because of miRNAs’ versatile and dynamic features. More research is needed to clarify the expression and role of miRNAs in sepsis and promote the use of miRNAs for sepsis management. This study provides an extensive, current, and thorough analysis of the involvement of miRNAs in sepsis. Its purpose is to encourage future research in this area, as tiny miRNAs have the potential to be used for rapid diagnosis, prognosis, and treatment of sepsis.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 2","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143052639","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lycopene Ameliorates Polycystic Ovary Syndrome in Rats by Inhibiting Ovarian Ferroptosis Through Activation of the AMPK/Nrf2 Pathway
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-27 DOI: 10.1002/jbt.70158
Kexin Wang, Lin Wang, Chengyong Wu, Hongxiang Chen, Donghui Cai, Linglan Lu, Xuli Liu, Zhen Jiao

Lycopene (LYC) is an extremely powerful antioxidant with the potential to treat a range of diseases and to inhibit ferroptosis. This research aims to elucidate how LYC impacts polycystic ovarian syndrome (PCOS) and the action mechanisms. A PCOS rat model was constructed by injecting DHEA. Different doses of LYC were injected intraperitoneally in PCOS rats, the estrous cycle was recorded. The histopathological damage of ovary in PCOS rats was observed by HE staining, testosterone (T), estradiol (E2), luteinizing hormone (LH) and follicle stimulating hormone (FSH) levels were examined by ELISA kits. Transmission electron microscopy, prussian blue staining, biochemical kits to determine ferroptosis. Immunohistochemistry and Western blot to assess the levels of ferroptosis-related and AMPK/Nrf2 pathway-related proteins to explore whether LYC affects ferroptosis in PCOS through this pathway. PCOS rats had significantly higher body weights, ovarian weights and ovarian indices, and disorganized estrous cycles, which were dose-dependently ameliorated by LYC. In addition, LYC significantly ameliorated the histopathological damage of ovary in PCOS rats and restored the normal secretion of T, E2, LH, and FSH. LYC attenuates iron deposition in PCOS ovarian tissues, reduces iron and ROS levels, and inhibits ferroptosis. Notably, LYC activated the AMPK/Nrf2 pathway, and AMPK inhibitor intervention attenuated the therapeutic effect of LYC in PCOS rats, suggesting that LYC acts through the AMPK/Nrf2 pathway. LYC attenuates estrous cycle disruption, ameliorates pathological impairments, and inhibits ferroptosis in PCOS rats by modulating the AMPK/Nrf2 pathway.

番茄红素(LYC)是一种极强的抗氧化剂,具有治疗一系列疾病和抑制铁氧化的潜力。本研究旨在阐明番茄红素如何影响多囊卵巢综合征(PCOS)及其作用机制。通过注射 DHEA,构建了 PCOS 大鼠模型。给多囊卵巢综合征大鼠腹腔注射不同剂量的 LYC,记录发情周期。用 HE 染色法观察 PCOS 大鼠卵巢的组织病理学损伤,用 ELISA 试剂盒检测睾酮(T)、雌二醇(E2)、黄体生成素(LH)和促卵泡激素(FSH)的水平。透射电子显微镜、普鲁士蓝染色、生化试剂盒确定铁变态反应。免疫组化和 Western 印迹法评估与铁氧化相关的蛋白和 AMPK/Nrf2 通路相关蛋白的水平,以探讨 LYC 是否通过该通路影响多囊卵巢综合征大鼠的铁氧化。PCOS大鼠的体重、卵巢重量和卵巢指数明显升高,发情周期紊乱,LYC对这些症状有剂量依赖性的改善作用。此外,LYC还能明显改善多囊卵巢综合征大鼠卵巢的组织病理学损伤,并恢复T、E2、LH和FSH的正常分泌。LYC 可减轻多囊卵巢综合征卵巢组织中的铁沉积,降低铁和 ROS 水平,抑制铁嗜酸性粒细胞增多。值得注意的是,LYC 激活了 AMPK/Nrf2 通路,AMPK 抑制剂的干预减弱了 LYC 对 PCOS 大鼠的治疗效果,这表明 LYC 是通过 AMPK/Nrf2 通路发挥作用的。LYC通过调节AMPK/Nrf2通路减轻了多囊卵巢综合征大鼠的发情周期紊乱、改善了病理损伤并抑制了铁蛋白沉积。
{"title":"Lycopene Ameliorates Polycystic Ovary Syndrome in Rats by Inhibiting Ovarian Ferroptosis Through Activation of the AMPK/Nrf2 Pathway","authors":"Kexin Wang,&nbsp;Lin Wang,&nbsp;Chengyong Wu,&nbsp;Hongxiang Chen,&nbsp;Donghui Cai,&nbsp;Linglan Lu,&nbsp;Xuli Liu,&nbsp;Zhen Jiao","doi":"10.1002/jbt.70158","DOIUrl":"10.1002/jbt.70158","url":null,"abstract":"<div>\u0000 \u0000 <p>Lycopene (LYC) is an extremely powerful antioxidant with the potential to treat a range of diseases and to inhibit ferroptosis. This research aims to elucidate how LYC impacts polycystic ovarian syndrome (PCOS) and the action mechanisms. A PCOS rat model was constructed by injecting DHEA. Different doses of LYC were injected intraperitoneally in PCOS rats, the estrous cycle was recorded. The histopathological damage of ovary in PCOS rats was observed by HE staining, testosterone (T), estradiol (E2), luteinizing hormone (LH) and follicle stimulating hormone (FSH) levels were examined by ELISA kits. Transmission electron microscopy, prussian blue staining, biochemical kits to determine ferroptosis. Immunohistochemistry and Western blot to assess the levels of ferroptosis-related and AMPK/Nrf2 pathway-related proteins to explore whether LYC affects ferroptosis in PCOS through this pathway. PCOS rats had significantly higher body weights, ovarian weights and ovarian indices, and disorganized estrous cycles, which were dose-dependently ameliorated by LYC. In addition, LYC significantly ameliorated the histopathological damage of ovary in PCOS rats and restored the normal secretion of T, E2, LH, and FSH. LYC attenuates iron deposition in PCOS ovarian tissues, reduces iron and ROS levels, and inhibits ferroptosis. Notably, LYC activated the AMPK/Nrf2 pathway, and AMPK inhibitor intervention attenuated the therapeutic effect of LYC in PCOS rats, suggesting that LYC acts through the AMPK/Nrf2 pathway. LYC attenuates estrous cycle disruption, ameliorates pathological impairments, and inhibits ferroptosis in PCOS rats by modulating the AMPK/Nrf2 pathway.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 2","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143052637","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sirtuin 1 Suppresses Hydrogen Peroxide-Induced Senescence and Promotes Viability and Migration in Lens Epithelial Cells by Inhibiting Forkhead Box Protein O1/Toll-Like Receptor 4 Pathway
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-27 DOI: 10.1002/jbt.70150
Hongda Jiang, Yuting Liu, Yinggui Yu, Yu Yan

Age-related cataracts (ARCs) are associated with increased oxidative stress and cellular senescence. Our objective is to investigate the function of Sirtuin 1 (SIRT1) within ARCs. In ARCs tissues and H2O2-treated lens epithelial cells (LECs), the expression levels of SIRT1 were examined. Senescence-associated β-galactosidase (SA-β-gal) staining was employed to evaluate cellular senescence. The Cell Counting Kit-8 assay was employed to measure viability. A wound healing assay was performed to assess migratory capacity in LECs. Oxidative stress-related indicators were determined by enzyme-linked immunosorbent assay kits. Additionally, the Coxpresdb and GeneCards databases were utilized to identify downstream pathways of SIRT1 in ARCs. The expression levels of protein and mRNA were detected using western blot and real-time quantitative polymerase chain reaction, respectively. The expression of SIRT1 was downregulated in ARCs tissues with an increase in reactive oxygen species. In H2O2-induced LECs, SIRT1 was downregulated and its overexpression inhibited oxidative stress and cellular senescence while promoting viability and migration. Furthermore, FoxO1/TLR4 pathway was screened out as the key pathway of SIRT1, which was activated in H2O2-induced LECs senescence. Overexpression of SIRT1 suppressed FoxO1/TLR4 pathway. Further research demonstrated that the activation of FoxO1/TLR4 pathway reversed the inhibitory role of SIRT1 in oxidative stress-induced cellular senescence and the promotion effect of SIRT1 on viability and migration in H2O2-induced LECs. SIRT1 inhibits oxidative stress-induced cellular senescence and promotes the viability and migration in H2O2-induced LECs via suppressing FoxO1/TLR4 pathway.

{"title":"Sirtuin 1 Suppresses Hydrogen Peroxide-Induced Senescence and Promotes Viability and Migration in Lens Epithelial Cells by Inhibiting Forkhead Box Protein O1/Toll-Like Receptor 4 Pathway","authors":"Hongda Jiang,&nbsp;Yuting Liu,&nbsp;Yinggui Yu,&nbsp;Yu Yan","doi":"10.1002/jbt.70150","DOIUrl":"10.1002/jbt.70150","url":null,"abstract":"<div>\u0000 \u0000 <p>Age-related cataracts (ARCs) are associated with increased oxidative stress and cellular senescence. Our objective is to investigate the function of Sirtuin 1 (SIRT1) within ARCs. In ARCs tissues and H<sub>2</sub>O<sub>2</sub>-treated lens epithelial cells (LECs), the expression levels of SIRT1 were examined. Senescence-associated β-galactosidase (SA-β-gal) staining was employed to evaluate cellular senescence. The Cell Counting Kit-8 assay was employed to measure viability. A wound healing assay was performed to assess migratory capacity in LECs. Oxidative stress-related indicators were determined by enzyme-linked immunosorbent assay kits. Additionally, the Coxpresdb and GeneCards databases were utilized to identify downstream pathways of SIRT1 in ARCs. The expression levels of protein and mRNA were detected using western blot and real-time quantitative polymerase chain reaction, respectively. The expression of SIRT1 was downregulated in ARCs tissues with an increase in reactive oxygen species. In H<sub>2</sub>O<sub>2</sub>-induced LECs, SIRT1 was downregulated and its overexpression inhibited oxidative stress and cellular senescence while promoting viability and migration. Furthermore, FoxO1/TLR4 pathway was screened out as the key pathway of SIRT1, which was activated in H<sub>2</sub>O<sub>2</sub>-induced LECs senescence. Overexpression of SIRT1 suppressed FoxO1/TLR4 pathway. Further research demonstrated that the activation of FoxO1/TLR4 pathway reversed the inhibitory role of SIRT1 in oxidative stress-induced cellular senescence and the promotion effect of SIRT1 on viability and migration in H<sub>2</sub>O<sub>2</sub>-induced LECs. SIRT1 inhibits oxidative stress-induced cellular senescence and promotes the viability and migration in H<sub>2</sub>O<sub>2</sub>-induced LECs via suppressing FoxO1/TLR4 pathway.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 2","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143046729","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Rutin Attenuates Pirarubicin-Induced Cardiomyocyte Injury by Knocking Down lncRNA Miat
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-26 DOI: 10.1002/jbt.70144
Peng Huang, Kaixu Yan, Wenqing Zhang, Jiahua Ji, Dexian Wei, Wentao Zheng, Lu Li, Liqun Ren

Pirarubicin (THP) is a widely used chemotherapeutic agent for breast cancer, but its clinical use is limited by its cardiotoxicity. In our previous study, we found that rutin (RUT), the main active ingredient in the traditional Chinese medicine Sophora japonica, could attenuate THP-induced cardiotoxicity. The aim of this study was to further investigate the potential role and mechanism of RUT in attenuating THP-induced cardiotoxicity. We examined the expression of lncRNA Miat in mouse cardiomyocytes (HL-1) under the intervention of THP/THP + RUT, respectively. Then we evaluated the protective effect of RUT on THP-induced HL-1 by CCK8, TUNEL, reactive oxygen species (ROS) assay and Western Blot. The mediating role of lncRNA Miat was verified by transfection of overexpression lncRNA Miat plasmid. The results showed that RUT increased cell viability, inhibited apoptosis, reduced ROS accumulation and decreased calcium overload in THP-induced HL-1. While overexpression of Miat significantly abrogated the therapeutic effect of rutin. In conclusion, RUT attenuates THP-induced myocardial injury by downregulating lncRNA Miat.

{"title":"Rutin Attenuates Pirarubicin-Induced Cardiomyocyte Injury by Knocking Down lncRNA Miat","authors":"Peng Huang,&nbsp;Kaixu Yan,&nbsp;Wenqing Zhang,&nbsp;Jiahua Ji,&nbsp;Dexian Wei,&nbsp;Wentao Zheng,&nbsp;Lu Li,&nbsp;Liqun Ren","doi":"10.1002/jbt.70144","DOIUrl":"https://doi.org/10.1002/jbt.70144","url":null,"abstract":"<div>\u0000 \u0000 <p>Pirarubicin (THP) is a widely used chemotherapeutic agent for breast cancer, but its clinical use is limited by its cardiotoxicity. In our previous study, we found that rutin (RUT), the main active ingredient in the traditional Chinese medicine Sophora japonica, could attenuate THP-induced cardiotoxicity. The aim of this study was to further investigate the potential role and mechanism of RUT in attenuating THP-induced cardiotoxicity. We examined the expression of lncRNA Miat in mouse cardiomyocytes (HL-1) under the intervention of THP/THP + RUT, respectively. Then we evaluated the protective effect of RUT on THP-induced HL-1 by CCK8, TUNEL, reactive oxygen species (ROS) assay and Western Blot. The mediating role of lncRNA Miat was verified by transfection of overexpression lncRNA Miat plasmid. The results showed that RUT increased cell viability, inhibited apoptosis, reduced ROS accumulation and decreased calcium overload in THP-induced HL-1. While overexpression of Miat significantly abrogated the therapeutic effect of rutin. In conclusion, RUT attenuates THP-induced myocardial injury by downregulating lncRNA Miat.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 2","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143119366","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Protective Effect of Fisetin on Subacute and Chronic Arsenic and Fluoride Co-Exposure Induced Hepatic, Renal and Cardiac Toxicities in Mice
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-26 DOI: 10.1002/jbt.70113
Sudipta Nayak, Debarati Rakshit, Vikas Yadav, Snehashis Kundu, Mohit Nema, Nilotpal Saharia, Awanish Mishra

Arsenic and fluoride are potent toxicants frequently present in drinking water and food, posing severe health risks through toxic impacts on key organs, primarily by inducing oxidative stress. Combined exposure to these elements exacerbates oxidative damage, yet effective strategies for preventing and managing their toxicities remain elusive. Fisetin, a bioflavonoid known for its antioxidant and anti-inflammatory properties, has demonstrated protective effects in multiple toxicity studies. However, its effect on arsenic- and fluoride-induced toxicity in vital organs (particularly the liver, kidneys, and heart) remains unexplored. This study investigates the protective potential of fisetin against subacute and chronic co-exposure to arsenic and fluoride-induced organ toxicity in a murine model. Male mice were administered arsenic and fluoride through drinking water, while fisetin (5, 10, and 20 mg/kg/day, orally) was co-administered simultaneously. Animals were sacrificed at 4 weeks (subacute) and 24 weeks (chronic) for liver, kidney, and heart tissue analysis, assessing biochemical markers and arsenic accumulation. Spectrophotometry study revealed some degree of arsenic chelation with fisetin. The results of In Vivo studies showed that both subacute and chronic exposure led to significant increases in pro-oxidant levels (ROS, TBARS, and nitrite) and arsenic accumulation, along with reduced GSH content in these organs compared to control. Fisetin treatment dose-dependently reduced oxidative stress and arsenic content in vital organs of exposed animals. The findings highlight fisetin's antioxidant activity and its capacity to reduce arsenic burden as potential protective mechanisms. These results support fisetin or fisetin-rich fruit consumption as a therapeutic strategy to mitigate organ toxicity from arsenic and fluoride co-exposure.

{"title":"Protective Effect of Fisetin on Subacute and Chronic Arsenic and Fluoride Co-Exposure Induced Hepatic, Renal and Cardiac Toxicities in Mice","authors":"Sudipta Nayak,&nbsp;Debarati Rakshit,&nbsp;Vikas Yadav,&nbsp;Snehashis Kundu,&nbsp;Mohit Nema,&nbsp;Nilotpal Saharia,&nbsp;Awanish Mishra","doi":"10.1002/jbt.70113","DOIUrl":"https://doi.org/10.1002/jbt.70113","url":null,"abstract":"<div>\u0000 \u0000 <p>Arsenic and fluoride are potent toxicants frequently present in drinking water and food, posing severe health risks through toxic impacts on key organs, primarily by inducing oxidative stress. Combined exposure to these elements exacerbates oxidative damage, yet effective strategies for preventing and managing their toxicities remain elusive. Fisetin, a bioflavonoid known for its antioxidant and anti-inflammatory properties, has demonstrated protective effects in multiple toxicity studies. However, its effect on arsenic- and fluoride-induced toxicity in vital organs (particularly the liver, kidneys, and heart) remains unexplored. This study investigates the protective potential of fisetin against subacute and chronic co-exposure to arsenic and fluoride-induced organ toxicity in a murine model. Male mice were administered arsenic and fluoride through drinking water, while fisetin (5, 10, and 20 mg/kg/day, orally) was co-administered simultaneously. Animals were sacrificed at 4 weeks (subacute) and 24 weeks (chronic) for liver, kidney, and heart tissue analysis, assessing biochemical markers and arsenic accumulation. Spectrophotometry study revealed some degree of arsenic chelation with fisetin. The results of In Vivo studies showed that both subacute and chronic exposure led to significant increases in pro-oxidant levels (ROS, TBARS, and nitrite) and arsenic accumulation, along with reduced GSH content in these organs compared to control. Fisetin treatment dose-dependently reduced oxidative stress and arsenic content in vital organs of exposed animals. The findings highlight fisetin's antioxidant activity and its capacity to reduce arsenic burden as potential protective mechanisms. These results support fisetin or fisetin-rich fruit consumption as a therapeutic strategy to mitigate organ toxicity from arsenic and fluoride co-exposure.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 2","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143119365","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Protective Role of Oxycodone in Myocardial Oxidative Stress and Mitochondrial Dysfunction Induced by Ischemia-Reperfusion
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-26 DOI: 10.1002/jbt.70151
Yongzheng Jiang, Hua He, Xinwei Jia

Ischemia-reperfusion (I/R) injury is a significant clinical problem impacting the heart and other organs, such as the kidneys and liver. This study explores the protective effects of oxycodone on myocardial I/R injury and its underlying mechanisms. Using a myocardial I/R model in Sprague-Dawley (SD) rats and an oxygen-glucose deprivation/reoxygenation (OGD/R) model in H9c2 cells, we administered oxycodone and inhibited AMP-activated protein kinase (AMPK) with Compound C (C.C). Our results showed that oxycodone significantly reduced lactate dehydrogenase (LDH) release and reactive oxygen species (ROS) production while stabilizing mitochondrial membrane potential (MMP). Western blot and RT-qPCR analyzes confirmed that oxycodone enhances AMPK phosphorylation and upregulates the expression of Silent Information Regulator 1 (SIRT1) and Peroxisome Proliferator-Activated Receptor γ Coactivator 1α (PGC-1α), thereby protecting myocardial cells. These findings suggest that oxycodone exerts significant protective effects against I/R injury by activating the AMPK pathway, offering new potential therapeutic targets for myocardial protection.

{"title":"Protective Role of Oxycodone in Myocardial Oxidative Stress and Mitochondrial Dysfunction Induced by Ischemia-Reperfusion","authors":"Yongzheng Jiang,&nbsp;Hua He,&nbsp;Xinwei Jia","doi":"10.1002/jbt.70151","DOIUrl":"10.1002/jbt.70151","url":null,"abstract":"<p>Ischemia-reperfusion (I/R) injury is a significant clinical problem impacting the heart and other organs, such as the kidneys and liver. This study explores the protective effects of oxycodone on myocardial I/R injury and its underlying mechanisms. Using a myocardial I/R model in Sprague-Dawley (SD) rats and an oxygen-glucose deprivation/reoxygenation (OGD/R) model in H9c2 cells, we administered oxycodone and inhibited AMP-activated protein kinase (AMPK) with Compound C (C.C). Our results showed that oxycodone significantly reduced lactate dehydrogenase (LDH) release and reactive oxygen species (ROS) production while stabilizing mitochondrial membrane potential (MMP). Western blot and RT-qPCR analyzes confirmed that oxycodone enhances AMPK phosphorylation and upregulates the expression of Silent Information Regulator 1 (SIRT1) and Peroxisome Proliferator-Activated Receptor γ Coactivator 1α (PGC-1α), thereby protecting myocardial cells. These findings suggest that oxycodone exerts significant protective effects against I/R injury by activating the AMPK pathway, offering new potential therapeutic targets for myocardial protection.</p>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 2","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/jbt.70151","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143046548","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mechanistic Insight of Pharmacological Aspects of Violacein: Recent Trends and Advancements
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-26 DOI: 10.1002/jbt.70114
Abhishek Chauhan, Darin Mansor Mathkor, Hemant Joshi, Ritu Chauhan, Ujjawal Sharma, Vikas Sharma, Manoj Kumar, Reena V. Saini, Adesh K. Saini, Hardeep Singh Tuli, Damandeep Kaur, Shafiul Haque

Since its discovery in the bacterium Chromobacterium violaceum, violacein—a striking purple pigment—has garnered significant interest due to its promising applications in the food and pharmaceutical industries. Violacein exhibits a range of pharmacological properties, including anti-inflammatory, anticancer, antibacterial, and antiparasitic effects, yet its complete molecular mechanisms are still being elucidated. Its mechanisms of action likely involve complex interactions with cellular receptors, signaling pathways, and specific molecular targets. Given violacein's unique properties and bioactive intermediates, future research holds substantial potential to advance its clinical and industrial applications. Upcoming studies will focus on deepening our understanding of violacein's molecular interactions, conducting clinical trials, and refining drug delivery systems to maximize its therapeutic value. Additionally, obtaining regulatory approval, conducting rigorous safety assessments, and developing efficient biosynthetic methods remain essential steps for violacein's successful integration into food biotechnology and medical applications.

{"title":"Mechanistic Insight of Pharmacological Aspects of Violacein: Recent Trends and Advancements","authors":"Abhishek Chauhan,&nbsp;Darin Mansor Mathkor,&nbsp;Hemant Joshi,&nbsp;Ritu Chauhan,&nbsp;Ujjawal Sharma,&nbsp;Vikas Sharma,&nbsp;Manoj Kumar,&nbsp;Reena V. Saini,&nbsp;Adesh K. Saini,&nbsp;Hardeep Singh Tuli,&nbsp;Damandeep Kaur,&nbsp;Shafiul Haque","doi":"10.1002/jbt.70114","DOIUrl":"10.1002/jbt.70114","url":null,"abstract":"<div>\u0000 \u0000 <p>Since its discovery in the bacterium <i>Chromobacterium violaceum</i>, violacein—a striking purple pigment—has garnered significant interest due to its promising applications in the food and pharmaceutical industries. Violacein exhibits a range of pharmacological properties, including anti-inflammatory, anticancer, antibacterial, and antiparasitic effects, yet its complete molecular mechanisms are still being elucidated. Its mechanisms of action likely involve complex interactions with cellular receptors, signaling pathways, and specific molecular targets. Given violacein's unique properties and bioactive intermediates, future research holds substantial potential to advance its clinical and industrial applications. Upcoming studies will focus on deepening our understanding of violacein's molecular interactions, conducting clinical trials, and refining drug delivery systems to maximize its therapeutic value. Additionally, obtaining regulatory approval, conducting rigorous safety assessments, and developing efficient biosynthetic methods remain essential steps for violacein's successful integration into food biotechnology and medical applications.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 2","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143046479","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Role of NLRP3 Inflammasome in Chronic Pain and Alzheimer's Disease—A Review
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-23 DOI: 10.1002/jbt.70071
Fatemeh Moradi, Tahmineh Mokhtari

The coexistence of Alzheimer's disease (AD) and chronic pain (CP) in the elderly population has been extensively documented, and a growing body of evidence supports the potential interconnections between these two conditions. This comprehensive review explores the mechanisms by which CP may contribute to the development and progression of AD, with a particular focus on neuroinflammatory pathways and the role of microglia, as well as the activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome. The review proposes that prolonged pain processing in critical brain regions can dysregulate the activity of the NLRP3 inflammasome within microglia, leading to the overproduction of pro-inflammatory cytokines and excessive oxidative stress in these regions. This aberrant microglial response also results in localized neuroinflammation in brain areas crucial for cognitive function. Additionally, CP as a persistent physiological and psychological stressor may be associated with hypothalamic-pituitary-adrenal (HPA) axis dysfunction, systemic inflammation, disruption of the blood–brain barrier (BBB), and neuroinflammation. These pathophysiological changes can cause morphological and functional impairments in brain regions responsible for cognition, memory, and neurotransmitter production, potentially contributing to the development and progression of CP-associated AD. Resultant neuroinflammation can further promote amyloid-beta (Aβ) plaque deposition, a hallmark of AD pathology. Potential therapeutic interventions targeting these neuroinflammatory pathways, particularly through the regulation of microglial NLRP3 activation, hold promise for improving outcomes in individuals with comorbid CP and AD. However, further research is required to fully elucidate the complex interplay between these conditions and develop effective treatment strategies.

{"title":"Role of NLRP3 Inflammasome in Chronic Pain and Alzheimer's Disease—A Review","authors":"Fatemeh Moradi,&nbsp;Tahmineh Mokhtari","doi":"10.1002/jbt.70071","DOIUrl":"10.1002/jbt.70071","url":null,"abstract":"<div>\u0000 \u0000 <p>The coexistence of Alzheimer's disease (AD) and chronic pain (CP) in the elderly population has been extensively documented, and a growing body of evidence supports the potential interconnections between these two conditions. This comprehensive review explores the mechanisms by which CP may contribute to the development and progression of AD, with a particular focus on neuroinflammatory pathways and the role of microglia, as well as the activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome. The review proposes that prolonged pain processing in critical brain regions can dysregulate the activity of the NLRP3 inflammasome within microglia, leading to the overproduction of pro-inflammatory cytokines and excessive oxidative stress in these regions. This aberrant microglial response also results in localized neuroinflammation in brain areas crucial for cognitive function. Additionally, CP as a persistent physiological and psychological stressor may be associated with hypothalamic-pituitary-adrenal (HPA) axis dysfunction, systemic inflammation, disruption of the blood–brain barrier (BBB), and neuroinflammation. These pathophysiological changes can cause morphological and functional impairments in brain regions responsible for cognition, memory, and neurotransmitter production, potentially contributing to the development and progression of CP-associated AD. Resultant neuroinflammation can further promote amyloid-beta (Aβ) plaque deposition, a hallmark of AD pathology. Potential therapeutic interventions targeting these neuroinflammatory pathways, particularly through the regulation of microglial NLRP3 activation, hold promise for improving outcomes in individuals with comorbid CP and AD. However, further research is required to fully elucidate the complex interplay between these conditions and develop effective treatment strategies.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 2","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143032984","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Biochemical and Molecular Toxicology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1